EP3864009A1 - Tyk2 kinase inhibitors - Google Patents

Tyk2 kinase inhibitors

Info

Publication number
EP3864009A1
EP3864009A1 EP19794095.0A EP19794095A EP3864009A1 EP 3864009 A1 EP3864009 A1 EP 3864009A1 EP 19794095 A EP19794095 A EP 19794095A EP 3864009 A1 EP3864009 A1 EP 3864009A1
Authority
EP
European Patent Office
Prior art keywords
compound
tyk2
compounds
disease
kinase
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP19794095.0A
Other languages
German (de)
English (en)
French (fr)
Inventor
John Charles Reader
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sareum Ltd
Original Assignee
Sareum Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sareum Ltd filed Critical Sareum Ltd
Publication of EP3864009A1 publication Critical patent/EP3864009A1/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D263/00Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings
    • C07D263/02Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings
    • C07D263/30Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D263/34Heterocyclic compounds containing 1,3-oxazole or hydrogenated 1,3-oxazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D263/48Nitrogen atoms not forming part of a nitro radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/541Non-condensed thiazines containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders

Definitions

  • This invention relates to compounds having Janus kinase inhibiting activity, and in particular TYK2 kinase inhibiting activity, pharmaceutical compositions containing them and their use in the treatment of various diseases such as autoimmune diseases.
  • Protein kinases constitute a large family of structurally related enzymes that are responsible for the control of a wide variety of signal transduction processes within the cell (Hardie and Hanks (1995) The Protein Kinase Facts Book. I and II, Academic Press, San Diego, CA).
  • the kinases may be categorized into families by the substrates they phosphorylate (e.g., protein-tyrosine, protein-serine/threonine, lipids, etc.). Sequence motifs have been identified that generally correspond to each of these kinase families (e.g., Hanks and Hunter, FASEB J., (1995) 9.
  • Protein kinases may be characterized by their regulation mechanisms.
  • An individual protein kinase may be regulated by more than one
  • phosphate groups processes, by adding phosphate groups to target proteins. These phosphorylation events act as molecular on/off switches that can modulate or regulate the target protein biological function. Phosphorylation of target proteins occurs in response to a variety of extracellular signals (hormones, neurotransmitters, growth and differentiation factors, etc.), cell cycle events, environmental or nutritional stresses, etc.
  • the appropriate protein kinase functions in signalling pathways to activate or inactivate (either directly or indirectly), for example, a metabolic enzyme, regulatory protein, receptor, cytoskeletal protein, ion channel or pump, or transcription factor.
  • JAK Janus kinase
  • JAK3 a family of intracellular non-receptor tyrosine kinases, ranging in size from 120-140 kDa, that transduce cytokine-mediated signals via the JAK- STAT pathway.
  • JAK family plays a role in the cytokine-dependent regulation of proliferation and function of cells involved in immune response.
  • JAK1 JAK1
  • JAK2 JAK3
  • TYK2 TYK2
  • JAK1 , JAK2 and TYK2 are ubiquitously expressed whereas JAK3 is expressed in the myeloid and lymphoid lineages.
  • the JAK family members are non-receptor tyrosine kinases that associate with many hematopoietin cytokines, receptor tyrosine kinases and GPCR's.
  • Each JAK kinase protein has a kinase domain and a catalytically inactive pseudo-kinase domain.
  • the JAK proteins bind to cytokine receptors through their amino-terminal FERM (Band-4.1 , ezrin, radixin, moesin) domains. After the binding of cytokines to their receptors, JAKs are activated and phosphorylate the receptors, thereby creating docking sites for signalling molecules, especially for members of the signal transducer and activator of transcription (STAT) family (Yamaoka et al, 2004. The Janus kinases (Jaks). Genome Biology 5(12): 253).
  • JAK1 , JAK2 and TYK2 are ubiquitously expressed.
  • TYK2 activates signal transducer and activator of transcription (STAT)-dependent gene expression and functional responses of interleukin-12, interleukin-23, and type I and III interferon receptors (Papp et al., The New England Journal of Medicine, 12 September 2018, DOI: 10.1056/NEJMoa1806382 and references cited therein)
  • STAT signal transducer and activator of transcription
  • interleukin-12, interleukin-23, and type I and III interferon receptors Papp et al., The New England Journal of Medicine, 12 September 2018, DOI: 10.1056/NEJMoa1806382 and references cited therein
  • These cytokine pathways are involved in the pathologic processes associated with immune-mediated disorders, including psoriasis, and are reported (Papp et al., idem) to be distinct from responses driven by Janus kinase (JAK) 1
  • Interleukin-23 composed of two subunits p19 and p40, is considered to be essential for the survival and expansion of Th17 cells which produce pro-inflammatory cytokines such as IL-17A, IL-17F, IL-6 and TNFa (see WO2014/07466 and references cited therein). These cytokines are reported as being critical in mediating the
  • autoimmune diseases including rheumatoid arthritis, multiple sclerosis, inflammatory bowel disease, and lupus.
  • IL-23 acts through a heterodimeric receptor composed of IL-12Rpi and IL-23R.
  • IL-12 in addition to the p40 subunit in common with IL-23, contains a p35 subunit and acts through a heterodimeric receptor composed of I L-12R1 b and I L-12R32.
  • IL-12 is essential for Th1 cell development and secretion of IFNy, a cytokine which plays a critical role in immunity by stimulating MHC expression, class switching of B cells to IgG subclasses, and the activation of macrophages (Gracie, J. A. et al., "Interleukin- 12 induces interferon- gamma-dependent switching of IgG alloantibody subclass", Eur. J. Immunol, 26: 1217- 1221 (1996); Schroder, K. et al., "Interferon-gamma: an overview of signals, mechanisms and functions", J. Leukoc. Biol, 75(2): 163-189 (2004)).
  • TYK2 associates with the IL-12Rpi subunit in the IL-12 and IL-23 receptors.
  • mice deficient in either p40, p19, or IL-23R are protected from disease in models of multiple sclerosis, rheumatoid arthritis, inflammatory bowel disease, lupus and psoriasis, among others (Kyttaris, V.C. et al, "Cutting edge: IL- 23 receptor deficiency prevents the development of lupus nephritis in C57BL/6-lpr/lpr mice", J. Immunol, 184:4605-4609 (2010); Hong, K.
  • Interleukin-23 rather than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain", Nature, 421 :744-748 (2003); Murphy, C.A. et al., "Divergent pro- and anti-inflammatory roles for IL-23 and IL-12 in joint autoimmune inflammation", J. Exp. Med, 198: 1951 -1957 (2003)).
  • TYK2 The role of TYK2 in the biological response to cytokines has been characterized using a mutant human cell line that was resistant to the effects of Type I interferons (IFNs) and by demonstrating that IFNa responsiveness could be restored by genetic complementation of TYK2 (Velazquez et al, 1992. Cell 70, 313-322). Further in vitro studies have implicated TYK2 in the signalling pathways of multiple other cytokines involved in both innate and adaptive immunity. However, analysis of TYK2 /_ mice revealed less profound immunological defects than were anticipated (Karaghiosoff et al, 2000. Immunity 13, 549- 560; Shimoda et al, 2000. Immunity 13, 561 -671 ).
  • TYK2 deficient mice display merely reduced responsiveness to IFNa/b and signal normally to interleukin 6 (IL- 6) and interleukin 10 (I L- 10), both of which activate TYK2 in vitro.
  • IL-6 interleukin 6
  • I L- 10 interleukin 10
  • TYK2 was shown to be essential for IL-12 signalling with the absence of TYK2 resulting in defective STAT4 activation and the failure of T cells from these mice to differentiate into IFNy- producing Th1 cells.
  • TYK2 /_ mice were more susceptible to viral and bacterial infections. The first patient with an autosomal recessive TYK2 deficiency was described by
  • cytokine signalling defects could be reponsible for many of the clinical manifestations described, for example atopic dermatitis and elevated IgE levels (enhanced Th2), increased incidence of viral infections (IFN defect), infection with intracellular bacteria (IL-12/Thl defect) and extracellular bacteria (IL-6 and I L-23/Th 17 defect).
  • IFN defect increased incidence of viral infections
  • IL-12/Thl defect infection with intracellular bacteria
  • IL-6 and I L-23/Th 17 defect extracellular bacteria
  • SNPs single nucleotide polymorphisms
  • TYK2 variants are associated with protection against systemic lupus erythematosus (SLE) (TYK2 rs2304256 and rsl2720270, NASAdsson et al, 2005. Am. J. Hum. Genet. 76, 528-537; Graham et al, 2007. Rheumatology 46, 927-930; Hellquist et al, 2009. J. Rheumatol. 36, 1631-1638; Jarvinen et al, 2010. Exp. Dermatol. 19, 123-131 ) and multiple sclerosis (MS) (rs34536443, Ban et al, 2009. Eur. J. Hum. Genet.
  • SLE systemic lupus erythematosus
  • TYK2 kinase Overexpression of TYK2 kinase has been implicated in the development of some disease states. For example, elevated levels of TYK2 were found in patients suffering from progressive pulmonary sarcoidosis (Schischmanoff et al., Sarcoidosis Vase. Diffuse., 2006, 23(2), 101-7).
  • TYK2 plays essential roles in both innate and adaptive immunity.
  • a lack of TYK2 expression manifests in the attenuated signalling of multiple proinflammatory cytokines and a profound imbalance in T helper cell differentiation.
  • evidence from genetic association studies supports that TYK2 is a shared autoimmune disease susceptibility gene. Taken together, these reasons suggest TYK2 as a target for the treatment of inflammatory and auto-immune diseases.
  • JAK family inhibitors have been reported in the literature which may be useful in the medical field (Ghoreschi et al, 2009. Immunol Rev, 228:273-287). It has been proposed that a selective TYK2 inhibitor that inhibits TYK2 with greater potency than JAK2 may have advantageous therapeutic properties, because inhibition of JAK2 can cause anemia (Ghoreschi et al, 2009. Nature Immunol. 4, 356-360). Papp et al. (The New England Journal of Medicine, 12 September 2018, DOI:
  • 10.1056/NEJMoa1806382 disclose the results obtained in Phase II clinical trials of the oral selective TYK2 inhibitor BMS-986165 in treating psoriasis and concluded that the results indicated a therapeutic benefit.
  • WO2014/074661 (Bristol-Myers Squibb) discloses a class of pyridazine and triazine amides as TYK2 inhibitors that are useful in the modulation of IL-12 IL-23 and/or IFNa. It is suggested that the compounds will be useful in the treatment of various inflammatory and autoimmune diseases.
  • WO2016/027195 discloses a series of aminopyrimidinyl compounds having JAK kinase inhibiting activity, incliuding activity agaist TYK2 kinase.
  • WO2012/000970 discloses a series of triazolopyridines as TYK2 kinase inhibitors.
  • WO201 1/113802 discloses a series of imidazopyridines as TYK2 kinase inhibitors.
  • the properties of JAK kinases and their relevance as therapeutic targets are also disclosed in W02008/156726, WO2009/155156, WO2010/005841 and WO2010/01 1375, all in the name of Merck.
  • WO2010/055304 and EP2634185 disclose a family of substituted oxazole carboxamides for use in the prophylaxis or treatment of autoimmune diseases and in particular multiple sclerosis.
  • WO2010/055304 are described as being FLT3 kinase inhibitors.
  • the kinase inhibiting effect of oxazole carboxamides is also disclosed in International patent application W02008/139161 (Sareum).
  • WO2015/032423 discloses the use of a subset of oxazole carboxamide compounds as TYK2 kinase inhibitors.
  • the compounds are described as being useful in the treatment of inflammatory and immunological disorders such as autmoimmune diseases.
  • WO2018/073438 discloses the the use of a subset of oxazole carboxamide compounds having TYK2 kinase inhibitory activity for use in treating T-cell lymphoblastic leukemias and cancers (such as hematopoietic cancers) which depend on the Janus kinase TYK2 for cancer cell survival.
  • the present invention relates to a small group of oxazole carboxamides that have improved activity against and selectivity for TYK2 kinase and improved pharmacokinetic properties compared to compounds disclosed in WO2015/032423 and WO2018/073438 and in particular the abovementioned Compound A and Compound B.
  • the invention provides a compound having the formula (1):
  • the invention provides a compound according to any one of Embodiments 1.1 to 1.3 wherein the compound is in a non-salt form.
  • acid salts may be formed with strong acids such as
  • salts can be formed, they can be synthesized from the parent compound by conventional chemical methods such as methods described in
  • such salts can be prepared by reacting the free base form of the compound with the acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media such as ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are used.
  • nonaqueous media such as ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are used.
  • salts can be formed, they may be pharmaceutically acceptable salts, and examples of pharmaceutically acceptable salts are discussed in Berge et a!., 1977, "Pharmaceutically Acceptable Salts," J. Pharm. Sci., Vol. 66, pp. 1-19.
  • salts that are not pharmaceutically acceptable may also be prepared as intermediate forms which may then be converted into pharmaceutically acceptable salts.
  • Such non- pharmaceutically acceptable salts forms which may be useful, for example, in the purification or separation of the compounds of the invention, also form part of the invention.
  • the compounds for use according to the invention as defined in any one of Embodiments 1.1 to 1.4 may contain one or more isotopic substitutions, and a reference to a particular element includes within its scope all isotopes of the element.
  • a reference to hydrogen includes within its scope 1 H, 2 H (D), and 3 H (T).
  • references to carbon and oxygen include within their scope respectively 12 C, 13 C and 14 C and 16 0 and 18 0.
  • the isotopes may be radioactive or non-radioactive.
  • the compound according to any one of Embodiments 1.1 to 1.4 contains no radioactive isotopes. Such compounds are preferred for therapeutic use.
  • Embodiments 1.1 to 1.4 may contain one or more radioisotopes. Compounds containing such radioisotopes may be useful in a diagnostic context.
  • Preferred solvates are solvates formed by the incorporation into the solid state structure (e.g. crystal structure) of the compounds of the invention of molecules of a non-toxic pharmaceutically acceptable solvent (referred to below as the solvating solvent).
  • Solvates can be prepared by recrystallising the compounds of the invention with a solvent or mixture of solvents containing the solvating solvent. Whether or not a solvate has been formed in any given instance can be determined by subjecting crystals of the compound to analysis using well known and standard techniques such as thermogravimetric analysis (TGE), differential scanning calorimetry (DSC) and X-ray crystallography.
  • TGE thermogravimetric analysis
  • DSC differential scanning calorimetry
  • X-ray crystallography X-ray crystallography
  • the solvates can be stoichiometric or non-stoichiometric solvates.
  • Particularly preferred solvates are hydrates, and examples of hydrates include hemihydrates, monohydrates and dihydrates.
  • the invention provides:
  • the compound of the invention may be anhydrous. Therefore, in another embodiment (Embodiment 1.9), the compound as defined in any one of Embodiments 1.1 to 1.6 is in an anhydrous form.
  • Biological Activity Compounds of the formulae (1 ), (2) and (3) as defined in Embodiments 1.1 to 1.9 are potent and selective inhibitors of TYK2 kinase.
  • the TYK2 kinase-inhibiting activities of the compounds can be determined using the assays described in the Examples below.
  • the TYK2 kinase-inhibiting activities of the compounds can be made use of in various methods of treating diseases where TYK2 plays a part in the development or progression of the disease.
  • the various uses of the compounds typically involve bringing the compounds into contact with a TYK2 kinase.
  • the inhibition of the TYK2 kinase may take place either in vitro or in vivo.
  • the invention provides:
  • a method of inhibiting a TYK2 kinase comprises bringing into contact with the TYK2 kinase an effective TYK2 kinase-inhibiting amount of a compound as defined in any one of Embodiments 1.1 to 1.9.
  • the inhibition of TYK2 kinase preferably takes place in vivo as part of a therapeutic treatment of a disease or condition in which TYK2 kinase is implicated.
  • the compounds of the invention are selective TYK2 inhibitors and are considerably more active against TYK2 than JAK2 and JAK3 kinases.
  • the compounds have relatively poor activity against a wide range of other kinases and, in particular, kinases that are generally recognised as targets for anti-cancer therapy.
  • the compounds have relatively little activity against Chk1 kinase, Aurora kinases, PKB (Akt) kinase and cyclin dependent kinases (CDK kinases) which are involved in cell cycle progression.
  • a lack of activity against kinases typically considered to be anti-cancer targets is beneficial in compounds that may be used in chronic treatment of inflammatory and autoimmune diseases for example.
  • the compounds of the invention will be useful in treating at least some of the diseases and disorders discussed below, including inflammatory diseases or conditions, immunological diseases or conditions, autoimmune diseases, allergic diseases or disorders, transplant rejections (allograft transplant rejections); Graft-versus host disease; treating sepsis and septic shock.
  • an autoimmune disease is a disease which is at least partially provoked by an immune reaction of the body against its own components, for example proteins, lipids or DNA.
  • organ-specific autoimmune disorders are insulin- dependent diabetes (Type I) which affects the pancreas, Hashimoto's thyroiditis and Graves' disease which affect the thyroid gland, pernicious anemia which affects the stomach, Cushing's disease and Addison's disease which affect the adrenal glands, chronic active hepatitis which affects the liver; polycystic ovary syndrome (PCOS), coeliac disease, psoriasis, inflammatory bowel disease (IBD), lupus nephritis (an inflammation of the kidney) and ankylosing spondylitis.
  • PCOS polycystic ovary syndrome
  • coeliac disease psoriasis
  • IBD inflammatory bowel disease
  • lupus nephritis an inflammation of the kidney
  • ankylosing spondylitis
  • non-organ-specific autoimmune disorders are rheumatoid arthritis, multiple sclerosis, systemic lupus erythematosus, and myasthenia gravis.
  • Type I diabetes ensues from the selective aggression of autoreactive T-cells against insulin secreting beta-cells of the islets of Langerhans.
  • Other inflammatory or immune diseases and disorders, sufferers from which may benefit from treatment with the compounds of the invention include skin inflammation due to radiation exposure; asthma; allergic inflammation; chronic inflammation; an inflammatory ophthalmic disease; dry eye syndrome (DES, also known as
  • keratoconjunctivitis sicca or dysfunctional tear syndrome e.g. chronic progressive or relapsing forms of non-infectious uveitis
  • alopecia areata e.g. chronic progressive or relapsing forms of non-infectious uveitis
  • alopecia areata e.g. chronic progressive or relapsing forms of non-infectious uveitis
  • alopecia areata e.g. chronic progressive or relapsing forms of non-infectious uveitis
  • alopecia areata e.g. chronic progressive or relapsing forms of non-infectious uveitis
  • alopecia areata e.g. chronic progressive or relapsing forms of non-infectious uveitis
  • alopecia areata e.g. chronic progressive or relapsing forms of non-infectious uveitis
  • RA Rheumatoid arthritis
  • RA is a chronic progressive, debilitating inflammatory disease that affects approximately 1% of the world's population.
  • RA is a symmetric polyarticular arthritis that primarily affects the small joints of the hands and feet.
  • pannus In addition to inflammation in the synovium, the joint lining, the aggressive front of tissue called pannus invades and destroys local articular structures (Firestein 2003, Nature 423:356-361 ).
  • IBD Inflammatory bowel disease
  • Crohn's disease involves most frequently the terminal ileum and colon, is transmural and discontinuous.
  • ulcerative colitis the inflammation is continuous and limited to rectal and colonic mucosal layers.
  • definitive classification of Crohn's disease or ulcerative colitis cannot be made and are designated 'indeterminate colitis'.
  • Both diseases include extraintestinal inflammation of the skin, eyes, or joints. Neutrophil-induced injuries may be prevented by the use of neutrophil migration inhibitors (Asakura et al., 2007, World J. Gastroenterol.
  • Psoriasis is a chronic inflammatory dermatosis that affects approximately 2% of the population. It is characterized by red, scaly skin patches that are usually found on the scalp, elbows, and knees, and may be associated with severe arthritis. The lesions are caused by abnormal keratinocyte proliferation and infiltration of inflammatory cells into the dermis and epidermis (Schon et al, 2005, New Engl. J. Med. 352: 1899-1912).
  • SLE Systemic lupus erythematosus
  • T cell-mediated B-cell activation results in glomerulonephritis and renal failure.
  • Human SLE is characterized at early stages by the expansion of long-lasting autoreactive CD4+ memory cells (D'Cruz et al, 2007, Lancet 369(9561 ):587-596).
  • Transplant rejection includes, without limitation, acute and chronic allograft rejection following for example transplantation of kidney, heart, liver, lung, bone marrow, skin and cornea. It is known that T cells play a central role in the specific immune response of allograft rejection. Hyperacute, acute and chronic organ transplant rejection may be treated. Hyperacute rejection occurs within minutes of transplantation. Acute rejection generally occurs within six to twelve months of the transplant. Hyperacute and acute rejections are typically reversible where treated with immunosuppressant agents. Chronic rejection, characterized by gradual loss of organ function, is an ongoing concern for transplant recipients because it can occur any time after transplantation.
  • GVDH graft-versus-host disease
  • Pulmonary sarcoidosis is a relatively rare inflammatory disorder of unknown cause, but which has been shown to be associated with elevated levels of TYK2, and which typically develops in adults of 20 to 50 years of age. Pulmonary sarcoidosis is characterised by small lumps, or granulomas in the lungs, which generally heal and disappear on their own. However, for those granulomas that do not heal, the tissue can remain inflamed and become scarred, or fibrotic. Pulmonary sarcoidosis can develop into pulmonary fibrosis, which distorts the structure of the lungs and can interfere with breathing.
  • the invention provides:
  • a compound as defined in any one of Embodiments 1.1 to 1.9 for use in the treatment of a disease or condition wherein the disease or condition is selected from an autoimmune disease, an inflammatory disease or condition, an immunological disease or condition, an allergic disease or disorder, a transplant rejection and Graft-versus host disease; or for use in the treatment of sepsis or septic shock, wherein the disease or condition is susceptible to TYK2 inhibition.
  • a compound as defined in any one of Embodiments 1.1 to 1.9 for the manufacture of a medicament for the treatment of a disease or condition selected from an autoimmune disease, an inflammatory disease or condition, an immunological disease or condition, an allergic disease or disorder, a transplant rejection and Graft-versus host disease; or for use in the treatment of sepsis or septic shock, wherein the disease or condition is susceptible to TYK2 inhibition.
  • a method of treating an autoimmune disease in a subject in need thereof comprises administering to the subject an effective TYK2 inhibiting amount of a compound as defined in any one of Embodiments 1.1 to 1.9, so as to inhibit TYK2 kinase in the subject and thereby block or reduce the extent of an inflammatory process associated with the autoimmune disease.
  • a compound as defined in any one of Embodiments 1.1 to 1.9 for use in a method of treating an autoimmune disease in a subject in need thereof, which method comprises administering to the subject an effective TYK2 inhibiting amount of the said compound, so as to inhibit TYK2 kinase in the subject and thereby block or reduce the extent of an inflammatory process associated with the autoimmune disease.
  • a method of treating a disease or condition in a subject in need thereof, wherein the disease or condition is one which is characterized or caused (at least in part) by or associated with overexpression (elevated expression) of TYK2 kinase which method comprises administering to the subject an effective TYK2 inhibiting amount of a compound of any one of Embodiments 1.1 to 1.9.
  • a compound as defined in any one of Embodiments 1.1 to 1.9 for use in treating a disease or condition in a subject in need thereof, wherein the disease is one which is characterized or caused (at least in part) by or associated with overexpression (elevated expression) of TYK2 kinase.
  • Embodiment 2.6 A method according to Embodiment 2.6 wherein the disease or condition is multiple sclerosis.
  • the activity of the compounds of the invention as TYK2 inhibitors can be measured using the assay set forth in the examples below and the level of activity exhibited by a given compound can be defined in terms of the IC50 value.
  • the compounds of the present invention have IC50 values against TYK2 kinase of less than 5 nanomolar.
  • the compound wherein R 1 is hydrogen has an IC50 against TYK2 of 1.9 nanomolar
  • the compound wherein R 1 is fluorine (Compound (3)) has an IC50 against TYK2 of 4.7 nanomolar.
  • An advantage of compounds of the invention is that they exhibit selectivity for TYK2 kinase compared to other kinases of the JAK family.
  • the compound wherein R 1 is hydrogen has approximately 25-fold selectivity for TYK2 compared to JAK2 and 110-fold selectivity for TYK2 compared to JAK3.
  • the compound where R 1 is fluorine (Compound (3)) has approximately 32-fold selectivity for TYK2 compared to JAK2 and 164-fold selectivity for TYK2 compared to JAK3.
  • the suitability of the compounds for use in treating psoriasis can be determined by testing the effect of the compounds on imiquimod-induced psoriasis-like skin inflammation in mice: see for example Mori et ai, Kobe J. Med. Sci., Vol. 62, No. 4, pp. E79-E88, 2016; van der Fits et al., The Journal of Immunology, 2009; 182: 5836-5845; and Lin et al., PLOS ONE
  • imiquimod can be applied topically to mice (for example to an ear of a mouse) to induce psoriasis- like inflammation and scaling, and a comparison made between the levels of inflammation and scaling in mice (or areas of the body of mice) that have also been treated with a compound of the invention or a control containing no imiquimod.
  • 2,6-dichlorobenzoyl chloride (10) is reacted with aminomalononitrile (1 1 ) (e.g. the p-toluenesulfonate salt thereof) in a polar aprotic solvent such as N-methylpyrrolidone (NMP) to give the amino-oxazole nitrile (12).
  • NMP N-methylpyrrolidone
  • the reaction is typically conducted at an elevated temperature, for example in the range from 90 °C to 1 15 °C.
  • the amino-oxazole nitrile (12) is converted to the corresponding bromo-compound (13) by a metal-free Sandmeyer procedure using tertiary butyl nitrite as a diazotizing agent in the presence of a halogen-donating compound such as bromo-(trimethyl)silane in dibromomethane.
  • the reaction is typically carried out under a protective (e.g. nitrogen) atmosphere at a temperature of about 0 °C.
  • the bromo-compound (13) is reacted with the substituted aniline (14) in a Buchwald- Hartwig palladium catalysed amination procedure to give the cyano-intermediate (15).
  • the reaction makes use of a palladium(O) catalyst such as bis(dibenzylideneacetone)- palladium(O) (Pd(dba) 2 ) in a polar aprotic solvent such as dioxane in the presence of a suitable phosphine ligand such as 1 ,T-ferrocenediyl-bis(diphenyl-phosphine) (dppf) or (5- diphenyl-phosphanyl-9,9-dimethyl-xanthen-4-yl)-diphenyl-phosphane, and a base such as potassium carbonate or caesium carbonate.
  • the reaction is typically carried out at an elevated temperature (for example from 95-125 °C), for example in a sealed tube, using microwave heating
  • the cyano-intermediate (15) is hydrolysed under mild acidic conditions (for example using sulphuric acid at a temperature of around 0 °C) to give the compound of formula (1 ) ⁇
  • mild acidic conditions for example using sulphuric acid at a temperature of around 0 °C
  • the invention provides:
  • a palladium (0) catalyst such as Pd(dba)2, a phosphine ligand (such as DPPF), and a base (such as potassium carbonate.
  • compositions comprising at least one active compound of the invention together with one or more pharmaceutically acceptable excipients such as carriers, adjuvants, diluents, fillers, buffers, stabilisers, preservatives, lubricants, or other materials well known to those skilled in the art, and optionally other therapeutic or prophylactic agents.
  • pharmaceutically acceptable excipients such as carriers, adjuvants, diluents, fillers, buffers, stabilisers, preservatives, lubricants, or other materials well known to those skilled in the art, and optionally other therapeutic or prophylactic agents.
  • pharmaceutically acceptable refers to compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of a subject (e.g. human) without excessive toxicity, irritation, allergic response, or other problems or complication, commensurate with a reasonable benefit/risk ratio.
  • a subject e.g. human
  • Each excipient must also be “acceptable” in the sense of being compatible with the other ingredients of the
  • compositions can be in any form suitable for oral, parenteral, topical, intranasal, ophthalmic, otic, rectal, intra-vaginal, or transdermal administration.
  • compositions are intended for parenteral administration, they can be formulated for intravenous, intramuscular, intraperitoneal, subcutaneous administration or for direct delivery into a target organ or tissue by injection, infusion or other means of delivery.
  • Pharmaceutical dosage forms suitable for oral administration include tablets, capsules, caplets, pills, lozenges, syrups, solutions, powders, granules, elixirs and suspensions, sublingual tablets, wafers or patches and buccal patches.
  • compositions containing compounds of the formulae (1 ), (2) and (3), or their pharmaceutically acceptable salts can be formulated in accordance with known techniques, see for example, Remington’s Pharmaceutical Sciences, Mack Publishing Company, Easton, PA, USA.
  • tablet compositions can contain a unit dosage of active compound together with an inert diluent or carrier such as a sugar or sugar alcohol, eg; lactose, sucrose, sorbitol or mannitol; and/or a non-sugar derived diluent such as sodium carbonate, calcium phosphate, calcium carbonate, or a cellulose or derivative thereof such as methyl cellulose, ethyl cellulose, hydroxypropyl methyl cellulose, and starches such as corn starch. Tablets may also contain such standard ingredients as binding and granulating agents such as polyvinylpyrrolidone, disintegrants (e.g.
  • swellable crosslinked polymers such as crosslinked carboxymethylcellulose
  • lubricating agents e.g. stearates
  • preservatives e.g. parabens
  • antioxidants e.g. BHT
  • buffering agents for example phosphate or citrate buffers
  • effervescent agents such as citrate/bicarbonate mixtures.
  • Capsule formulations may be of the hard gelatin or soft gelatin variety and can contain the active component in solid, semi-solid, or liquid form.
  • Gelatin capsules can be formed from animal gelatin or synthetic or plant derived equivalents thereof.
  • the solid dosage forms can be coated or un-coated, but typically have a coating, for example a protective film coating (e.g. a wax or varnish) or a release controlling coating.
  • a protective film coating e.g. a wax or varnish
  • the coating e.g. a EudragitTM type polymer
  • the coating can be designed to release the active component at a desired location within the gastro-intestinal tract.
  • the coating can be selected so as to degrade under certain pH conditions within the gastrointestinal tract, thereby selectively releasing the compound in the stomach or in the ileum or duodenum.
  • the drug can be presented in a solid matrix comprising a release controlling agent, for example a release delaying agent which may be adapted to selectively release the compound under conditions of varying acidity or alkalinity in the gastrointestinal tract.
  • a release controlling agent for example a release delaying agent which may be adapted to selectively release the compound under conditions of varying acidity or alkalinity in the gastrointestinal tract.
  • the matrix material or release retarding coating can take the form of an erodible polymer (e.g. a maleic anhydride polymer) which is substantially continuously eroded as the dosage form passes through the gastrointestinal tract.
  • the active compound can be formulated in a delivery system that provides osmotic control of the release of the compound.
  • Osmotic release and other delayed release or sustained release formulations may be prepared in accordance with methods well known to those skilled in the art.
  • compositions for topical use include ointments, creams, sprays, patches, gels, liquid drops and inserts (for example intraocular inserts). Such compositions can be formulated in accordance with known methods.
  • compositions for parenteral administration are typically presented as sterile aqueous or oily solutions or fine suspensions, or may be provided in finely divided sterile powder form for making up extemporaneously with sterile water for injection.
  • compositions for parenteral administration may be formulated for administration as discrete dosage units or may be formulated for administration by infusion.
  • formulations for rectal or intra-vaginal administration include pessaries and suppositories which may be, for example, formed from a shaped mouldable or waxy material containing the active compound.
  • compositions for administration by inhalation may take the form of inhalable powder compositions or liquid or powder sprays, and can be administrated in standard form using powder inhaler devices or aerosol dispensing devices. Such devices are well known.
  • the powdered formulations typically comprise the active compound together with an inert solid powdered diluent such as lactose.
  • a formulation intended for oral administration may contain from 0.1 milligrams to 2 grams of active ingredient, more usually from 10 milligrams to 1 gram, for example, 50 milligrams to 500 milligrams.
  • the active compound will be administered to a patient in need thereof (for example a human or animal patient) in an amount sufficient to achieve the desired therapeutic effect.
  • the compounds will typically be administered in amounts that are therapeutically or prophylactically useful and which generally are non-toxic.
  • the benefits of administering a compound of the formula (1 ), (2) or (3) may outweigh the disadvantages of any toxic effects or side effects, in which case it may be considered desirable to administer compounds in amounts that are associated with a degree of toxicity.
  • the compounds may be administered over a prolonged term to maintain beneficial therapeutic effects or may be administered for a short period only. Alternatively they may be administered in a pulsatile or continuous manner.
  • the compound of formula (1 ), (2) or (3) will generally be administered to a subject in need of such administration, for example a human patient.
  • a typical daily dose of the compound can be up to 1000 mg per day, for example in the range from 0.01 milligrams to 10 milligrams per kilogram of body weight, more usually from 0.025 milligrams to 5 milligrams per kilogram of body weight, for example up to 3 milligrams per kilogram of bodyweight, and more typically 0.15 milligrams to 5 milligrams per kilogram of bodyweight although higher or lower doses may be administered where required.
  • an initial starting dose of 12.5 mg may be administered 2 to 3 times a day.
  • the dosage can be increased by 12.5 mg a day every 3 to 5 days until the maximal tolerated and effective dose is reached for the individual as determined by the physician.
  • the quantity of compound administered will be commensurate with the nature of the disease or physiological condition being treated and the therapeutic benefits and the presence or absence of side effects produced by a given dosage regimen, and will be at the discretion of the physician.
  • the compounds of the formulae (1 ), (2) and (3) can be administered as the sole therapeutic agent or they can be administered in combination therapy with one or more other compounds such as steroids, interferons, apremilast (for psoriasis) or methotrexate (for rheumatoid arthritis).
  • a patient Prior to administration of a compound of the invention, a patient may be screened to determine whether a disease or condition from which the patient is or may be suffering is one which would be susceptible to treatment with a compound having activity against TYK2.
  • the invention provides:
  • a method for the diagnosis and treatment of a disease state or condition mediated by TYK2 kinase comprises (i) screening a patient to determine whether a disease or condition from which the patient is or may be suffering is one which would be susceptible to treatment with a compound having activity against the kinase; and (ii) where it is indicated that the disease or condition from which the patient is thus susceptible, thereafter administering to the patient an effective TYK2 inhibiting amount of a compound of any one of Embodiments 1.1 to 1.9.
  • a subject e.g. patient
  • a diagnostic test to detect a marker indicative of the presence of a disease or condition in which TYK2 is implicated, or a marker indicative of susceptibility to the said disease or condition.
  • subjects may be screened for genetic markers indicative of a susceptibility to develop an autoimmune or inflammatory disease.
  • the genetic marker can comprise a particular allele or single nucleotide polymorphism of the TYK2 gene which is indicative of susceptibility to an autoimmune disease such as multiple sclerosis (see for example Ban et al., European Journal of Human Genetics (2009), 17, 1309-1313) or an inflammatory bowel disease such as Crohn’s disease (see Sato et al., J. Clin. Immunol. (2009), 29:815-825).
  • the genetic marker can, for example, be a single nucleotide polymorphism in the TYK2 gene, or it can be a haplotype comprising a single nucleotide polymorphism in the TYK2 gene and a polymorphism in another gene.
  • the diagnostic tests are typically conducted on a biological sample selected from blood samples, biopsy samples, stool biopsies, sputum, chromosome analysis, pleural fluid, peritoneal fluid, or urine.
  • the compounds of the invention were assayed for their ability to inhibit TYK2 kinase and other JAK kinases.
  • the activities of the compounds were compared with the activities of Compound A (2-(2-chloro-6-fluoro-phenyl)-5-[4-(morpholine-4-carbonyl)-phenylamino]- oxazole-4-carboxylic acid amide) and Compound B (2-(2,6-dichloro-phenyl)-5-[4- (morpholine-4-carbonyl)-phenylamino]-oxazole-4-carboxylic acid amide):
  • Kinase assays were performed at Reaction Biology Corp., Malvern, Pennsylvania, USA, using the general procedure set out below. In the assays, the ATP concentration was 100 mM and the top compound concentrations were 10 mM.
  • TYK2 and JAK kinase data in Table 7 on page 61 of WO 2015/032423 were generated using an assay in which the ATP concentration was 10 mM whereas, as indicated above, the assay described in the protocol below used an ATP concentration of 100 mM.
  • JAK3tide [Ac-GEEEEYFELVKKKK-NH 2 ]
  • Test compounds prepared and serially diluted in DMSO, were incubated at six concentrations (1 % DMSO final) with pooled human liver microsomes in the presence of probe substrate for each isoform, and their effects on the metabolism of probe substrates determined. Incubations (in 96-well plates) were carried out at 37 °C in 0.1 M Tris buffer, pH 7.4, with reactions initiated by the addition of cofactor, NADPH (1 mM final concentration).
  • the CYP450 isoforms studied, and their respective probe substrates are shown in Table 4.
  • the potential for compounds to inhibit the hERG potassium channel was determined using a hERG-HEK stably transfected cell line on the Sophion Qube automated electrophysiology platform.
  • the assay was performed at room temperature and recordings of the hERG tail current from individual cells was made using single-hole QChips.
  • the potency (I C50) of test compounds to inhibit the hERG channel was determined from a concentration-response curve generated from 8 test compound concentrations with up to 4 replicates per concentration.
  • the compound concentration was added to the test well twice to ensure complete exchange of the external buffer with the test compound. In total, compound was applied to the well for >7 minutes.
  • the compounds (2) and (3) of the invention and prior art comparative Compound B were tested in hepatocyte stability assays which were performed using pooled hepatocytes from mouse (male CD-1 ), rat (male Sprague-Dawley), dog (male Beagle) and human (mixed gender). Test and control compounds were incubated with hepatocytes at 37 °C. Aliquots were removed at 6 timepoints over a one hour period. Samples were centrifuged and the supernatant fractions analysed for parent compound by mass spectrometry (LC- MS/MS).
  • LC- MS/MS mass spectrometry
  • HT29 cells were serum starved overnight before the four test compounds were diluted to generate a 9-point semi-log dose dilution with a top concentration of 10 mM, plus vehicle control.
  • HT29 cells were incubated with the test compounds for 20 min at 37°C.
  • HT29 cells were incubated for a further 15 min with 10 ng/ml human IL-22 before cells were fixed with 4% PFA for 10 min, and 90% methanol for 30 min before being labelled with a phospho-STAT3Y705 antibody (CST #9145).
  • Cells were rinsed three times using 0.5% BSA/PBS solution before being incubated with Alexa-488 anti-rabbit secondary antibodies.
  • Fresh human Peripheral blood CD4CD45RO+ cells were purchased commercially (Generon, UK); 3 separate vials from 3 different volunteers for experimental replicates. Cells were grown in T-cell medium (Thermo Fisher) containing 10ng/ml recombinant human IL-1 B (R&D Systems), IL-23 (R&D Systems), TGF-B1 (R&D Systems) and
  • IL-6 50ng/ml IL-6 (R&D Systems) together with anti-CD3/CD28 magnetic Dynabeads (Thermo Fisher). These were grown for 1 1 days to induce expansion of Th17 cells. Prior to plating for assays cells were grown overnight in T-cell medium supplemented with human serum (1 %) overnight. Media was removed and replaced with unsupplemented RPMI for 4h prior to assay.
  • pSTAT3 levels in the lysates were measured using a commercially available ELISA kit (Thermo Fisher; 85- 86102-1 1 ).
  • ELISAs were carried out according to manufacturers instructions and absorbance read using a microplate reader (Thermo Fisher; Varioskan). Data was normalised to the response in untreated samples using the formula:
  • % of control ((Stimulated sample Cone. - unstimulated sample Cone.) X
  • both compounds (2) and (3) are more active than Compound B in the TYK2 kinase inhibition assay and both have greater selectivity for TYK2 versus JAK1 , JAK2 and JAK3 kinases than Compound B.
  • Compounds (2) and (3) have slightly advantageous properties compared to prior art comparative Compound B in the cytochrome P450 assays, notably in the CYP2C8 and CYP2C9 assays. Compounds (2) and (3) have a reduced hERG liability compared to prior art comparative Compound B.
  • Compounds (2) and (3) showed a reduced rate of clearance and a consequently longer half life than comparative Compound B.
  • Compound (2) is more potent in inhibiting phosphorylation of STAT3 in IL-22 stimulated HT29 cells and Th17 cells compared to comparative Compound B.
  • Compound (2) shows a greater inhibition of IL-17F production in Th17 cells compared to comparative Compound B.
  • a tablet composition containing a compound of the formula (2) or formula (3) or a pharmaceutically acceptable salt thereof is prepared by mixing 50mg of the compound with 197mg of lactose (BP) as diluent, and 3mg magnesium stearate as a lubricant and compressing to form a tablet in a known manner.
  • BP lactose
  • a capsule formulation is prepared by mixing 100mg of a compound of the formula (2) or formula (3) or a pharmaceutically acceptable salt thereof with 10Omg lactose and filling the resulting mixture into standard opaque hard gelatin capsules.
  • a composition for sub-cutaneous administration is prepared by mixing a compound of the formula (2) or formula (3) with pharmaceutical grade corn oil to give a concentration of 5mg/mL.
  • the composition is sterilised and filled into a suitable container.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Rheumatology (AREA)
  • Pain & Pain Management (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
EP19794095.0A 2018-10-08 2019-10-07 Tyk2 kinase inhibitors Pending EP3864009A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB1816369.1A GB201816369D0 (en) 2018-10-08 2018-10-08 Pharmaceutical compounds
PCT/EP2019/077118 WO2020074461A1 (en) 2018-10-08 2019-10-07 Tyk2 kinase inhibitors

Publications (1)

Publication Number Publication Date
EP3864009A1 true EP3864009A1 (en) 2021-08-18

Family

ID=64397425

Family Applications (1)

Application Number Title Priority Date Filing Date
EP19794095.0A Pending EP3864009A1 (en) 2018-10-08 2019-10-07 Tyk2 kinase inhibitors

Country Status (13)

Country Link
US (1) US20210387981A1 (zh)
EP (1) EP3864009A1 (zh)
JP (1) JP7383030B2 (zh)
KR (1) KR20210076036A (zh)
CN (1) CN113056456B (zh)
AU (1) AU2019359320B2 (zh)
BR (1) BR112021006424A2 (zh)
CA (1) CA3111049A1 (zh)
GB (1) GB201816369D0 (zh)
IL (1) IL281599B1 (zh)
MX (1) MX2021003929A (zh)
SG (1) SG11202102490TA (zh)
WO (1) WO2020074461A1 (zh)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB202005114D0 (en) * 2020-04-07 2020-05-20 Sareum Ltd Crystalline Forms of a Pharmaceutical Compound
CA3236262A1 (en) 2021-10-25 2023-05-04 Isaac Marx Tyk2 degraders and uses thereof

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0709031D0 (en) * 2007-05-10 2007-06-20 Sareum Ltd Pharmaceutical compounds
AU2008266883B2 (en) 2007-06-20 2014-01-30 Merck Sharp & Dohme Corp. Inhibitors of janus kinases
JP2011518221A (ja) 2008-04-21 2011-06-23 メルク・シャープ・エンド・ドーム・コーポレイション Janusキナーゼの阻害剤
AU2009260389A1 (en) 2008-06-18 2009-12-23 Merck Sharp & Dohme Corp. Inhibitors of Janus kinases
EP2310384B1 (en) 2008-07-09 2014-04-09 Merck Sharp & Dohme Corp. Inhibitors of janus kinases
GB0820819D0 (en) 2008-11-13 2008-12-24 Sareum Ltd Pharmaceutical compounds
MX2012010265A (es) 2010-03-17 2012-10-01 Hoffmann La Roche Compuestos de imidazopiridina, composiciones y metodos de uso.
WO2012000970A1 (en) 2010-07-01 2012-01-05 Cellzome Limited Triazolopyridines as tyk2 inhibitors
DK2634185T3 (en) * 2012-03-02 2016-03-21 Sareum Ltd Tyk2 kinase inhibitors
KR101202026B1 (ko) 2012-07-03 2012-11-16 민호현 황토 석부작 제조 방법
MX2015005731A (es) 2012-11-08 2015-09-16 Bristol Myers Squibb Co Compuestos heterciclicos sustituidos con amida utiles como moduladores de las respuestas de interleucina 12(il-12), interleucina 23 (il-23) y/o interferon alfa (ifnalfa).
CN105793245B (zh) * 2013-09-03 2018-03-20 萨勒姆有限公司 药物化合物
NO2721710T3 (zh) 2014-08-21 2018-03-31
GB201617871D0 (en) 2016-10-21 2016-12-07 Sareum Limited Pharmaceutical compounds

Also Published As

Publication number Publication date
AU2019359320A1 (en) 2021-03-18
SG11202102490TA (en) 2021-04-29
KR20210076036A (ko) 2021-06-23
CN113056456B (zh) 2023-06-13
IL281599A (en) 2021-05-31
WO2020074461A1 (en) 2020-04-16
MX2021003929A (es) 2021-06-04
US20210387981A1 (en) 2021-12-16
AU2019359320B2 (en) 2024-04-04
CN113056456A (zh) 2021-06-29
CA3111049A1 (en) 2020-04-16
BR112021006424A2 (pt) 2021-07-06
GB201816369D0 (en) 2018-11-28
JP2022514799A (ja) 2022-02-15
IL281599B1 (en) 2024-06-01
JP7383030B2 (ja) 2023-11-17

Similar Documents

Publication Publication Date Title
US11673870B2 (en) Pharmaceutical compounds
KR102191084B1 (ko) 약제학적 화합물
JP2019501125A (ja) IL−12、IL−23および/またはIFNα応答のモジュレーターとして有用なイミダゾピリダジン化合物
JP7012082B2 (ja) Il-12、il-23および/またはifn-アルファのイミダゾピリダジン調節剤
AU2019359320B2 (en) TYK2 kinase inhibitors
US8871786B2 (en) Azaindazole amide compounds as CCR1 receptor antagonists
US20230159473A1 (en) Crystalline forms of a tyk2 inhibitor
RU2805932C2 (ru) Фармацевтические соединения

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20210409

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40048146

Country of ref document: HK

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20220214

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20240314

GRAJ Information related to disapproval of communication of intention to grant by the applicant or resumption of examination proceedings by the epo deleted

Free format text: ORIGINAL CODE: EPIDOSDIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

INTC Intention to grant announced (deleted)
GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED